1
|
Zhang M, Zhong J, Song Z, Xu Q, Chen Y, Zhang Z. Regulatory mechanisms and potential therapeutic targets in precancerous lesions of gastric cancer: A comprehensive review. Biomed Pharmacother 2024; 177:117068. [PMID: 39018877 DOI: 10.1016/j.biopha.2024.117068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024] Open
Abstract
Precancerous lesions of gastric cancer (PLGC) represent a critical pathological stage in the transformation from normal gastric mucosa to gastric cancer (GC). The global incidence of PLGC has been rising over the past few decades, with a trend towards younger onset ages. Increasing evidence suggests that early prevention and treatment of PLGC can effectively reverse the malignant development of gastric mucosal epithelial cells. However, there is currently a lack of effective therapeutic drugs and methods. Recent years have witnessed substantial advancements in PLGC research, with the elucidation of novel regulatory mechanisms offering promising avenues for clinical intervention and drug development. This review aims to delineate potential targets for early prevention and diagnosis of GC while exploring innovative approaches to PLGC management. This article focuses on elucidating the regulatory mechanisms of the inflammatory microenvironment, bile acids (BA), glycolysis, autophagy, apoptosis, ferroptosis, and cellular senescence. We pay particular attention to potential therapeutic targets for PLGC, with the goal of providing insights and theoretical basis for clinical research on PLGC.
Collapse
Affiliation(s)
- Maofu Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Jialin Zhong
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhongyang Song
- Department of Oncology, Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730020, China
| | - Qian Xu
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yuchan Chen
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhiming Zhang
- Department of Oncology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu 730050, China.
| |
Collapse
|
2
|
Hnatyszyn A, Szalata M, Zielińska A, Wielgus K, Danielewski M, Hnatyszyn PT, Pławski A, Walkowiak J, Słomski R. Mutations in Helicobacter pylori infected patients with chronic gastritis, intestinal type of gastric cancer and familial gastric cancer. Hered Cancer Clin Pract 2024; 22:9. [PMID: 38867324 PMCID: PMC11167877 DOI: 10.1186/s13053-024-00282-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Development of sequential changes of mucous leading to gastric cancer and familial cases of gastric cancer of intestinal type is widely connected with Helicobacter pylori infections. In this study we analysed variants of genes involved in cancerogenesis and inflammatory processes of intestines in patients infected with H.pylori. Our goal was to test whether mutations in these genes predestinate to development of gastric cancer, and whether there is a genetic factor that makes it more likely for infections with H.pylori to cause gastric cancer. As infections with H. pylori are relatively common, discovering such genetic predispositions could be used for establishing risk-groups and for planning treatments. METHODS Our studies cover analysis of variants in genes involved in cancerogenesis: TP53 (rs11540652, rs587782329, COSM10771), MSH2 (rs193922376), MLH1 (rs63750217), and inflammatory processes of intestine: NOD2 (rs2066847, rs2066842), IL1A (rs1800587) and IL1B (rs1143634) from H.pylori-infected patients. RESULTS Mutations were more common in the group of patients with gastric cancer of intestinal type and familial cases of gastric cancer in comparison with patients with chronic gastritis, chronic atrophic gastritis, intestinal metaplasia, dysplasia or gastric cancer (p-value = 0.00824), with the prevalence of p53 mutations in patients with familial gastric cancer vs. patients with other changes of mucosa (p-value = 0.000049). Additionally, gastric cancer patients have mainly genotype TT or CT of the rs2066842 variant of the NOD2 gene. CONCLUSIONS The lack of statistically significant changes of other interleukin genes involved in inflammatory processes may suggest the presence of H.pylori infection as a potential trigger for the development of the inflammatory process of the mucosa, leading through microbiota dysbiosis to the development of enteric gastric cancer. Mutations in analysed genes correlated with more severe mucosal changes, with a much more frequent presence of TP53 gene mutations, with a limited presence of other mutations in the familial history of gastric cancer.
Collapse
Affiliation(s)
- Andrzej Hnatyszyn
- Independent Public Health Care Centre in Nowa Sol, Multispecialty Hospital, Chalubinskiego 7, Nowa Sol, 67-100, Poland
| | - Marlena Szalata
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Dojazd 11, Poznań, 60-632, Poland
| | - Aleksandra Zielińska
- Department of Biotechnology, Institute of Natural Fibres and Medicinal Plants National Research Institute, Wojska Polskiego 71B, Poznań, 60-630, Poland
| | - Karolina Wielgus
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Szpitalna 27/33, Poznań, 60-572, Poland
| | - Mikołaj Danielewski
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Szpitalna 27/33, Poznań, 60-572, Poland
| | | | - Andrzej Pławski
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, Poznań, 60-479, Poland
- Department of General, Endocrinological Surgery and Gastrointestinal Oncology, Institute of Surgery, Poznan University of Medical Sciences, Przybyszewskiego 49, Poznań, 60-355, Poland
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Szpitalna 27/33, Poznań, 60-572, Poland
| | - Ryszard Słomski
- Department of Biotechnology, Institute of Natural Fibres and Medicinal Plants National Research Institute, Wojska Polskiego 71B, Poznań, 60-630, Poland.
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, Poznań, 60-479, Poland.
| |
Collapse
|
3
|
Ibrahim A, Kamel NO, Rageh F, Elgamal R, Mansour Salama B, Sakr MA, Elhoseeny MM, Osman EM, Sayed S, Ramadan ME. The involvement of cytokine gene polymorphism in determining the vulnerability to Blastocystis and Helicobacter pylori co-infection in the Egyptian population. Acta Trop 2024; 252:107137. [PMID: 38364930 DOI: 10.1016/j.actatropica.2024.107137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/18/2024]
Abstract
AIMS The present study aimed to identify any potential association between IL-1β and TNF-α gene polymorphism and the risk of Blastocystis infection as well as co-infection of Blastocystis with Helicobacter pylori (H.pylori). METHODOLOGY A total of 314 stool samples were collected and examined microscopically for the detection of parasitic infection. DNA was extracted from all samples and utilized to identify Blastocystis molecularly. Positive samples were used for H. pylori detection by rapid tests and PCR. Moreover, we investigate polymorphism in the TNF-α gene at position -1031T/C, -308 G/A, and IL-1β at position +3954C/T using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assay. RESULTS Out of the 314 stool samples, Blastocystis was detected in 93 (29.6 %); among them, 54 (58.1 %) had a mixed infection of Blastocystis with H. pylori. The TT genotype of the IL-1β gene at position +3954 was significantly higher in Blasocystis-infected patients than in uninfected patients (17.2% vs. 6.3 %, P = 0.02), which might be considered a risk factor (OR = 3.2; CI =1.21-8.52). The TNF-α at position -1031 TT genotype was significantly higher in Blastocystis-infected patients than uninfected patients (44.1% vs. 10.8 %, P< 0.0001). The T allele (OR= 2.67; CI=1.51-4.72, P = 0.0008) might be considered a risk factor. The TNF- α at position -308 AA genotype is higher in Blasocystis infected than uninfected (17.2% vs 7.2 %, P = 0.03). TNF-α -308 AA (OR = 2.72; CI = 1.08-6.89) and A allele (OR= 1.46; CI= 0.797-2.66) might be considered risk factors. The TNF- α at position -308 G/A showed that the GG is the most frequent genotype in Blastocystis with H. pylori-positive patients with a significant association (P = 0.004), as well as the G allele (P = 0.02). The G allele (OR=1.924; CI= 1.071-3.454) might be considered a risk factor for co-infection of Blastocystis and H. pylori. CONCLUSION SNPs (-1031 T/C and -308 G/A) of the TNF-α and (+3954 C/T) of the IL-1β may be a useful marker in the assessment of the risk of Blastocystis infection, and TNF-α at position -308 G/A) may be a predictor for co-infection of Blastocystis with H. pylori.
Collapse
Affiliation(s)
- Asmaa Ibrahim
- Genetic engineering and Biotechnology research institute, University of Sadat City (GEBRI, USC), Egypt; Departments of Medical Parasitology, Faculty of Medicine, Cairo University (Laboratory of Molecular Medical Parasitology, LMMP), Egypt.
| | - Nancy O Kamel
- Department of Medical Parasitology, Faculty of Medicine, October 6 University, Egypt
| | - Fatma Rageh
- Infectious disease, Gastroenterology and hepatology department, Suez University, Egypt
| | - Rasha Elgamal
- Clinical pathology department, Faculty of Medicine, Suez University, Egypt
| | - Bassam Mansour Salama
- Infectious and Endemic Diseases Department, Faculty of Medicine Suez Canal University, Egypt
| | - Mohamed A Sakr
- Medical Microbiology and Immunology Department, Faculty of Medicine, Suez University, P.O. Box:43221, Suez, Egypt
| | | | - Eman M Osman
- Immunology and allergy department, Medical Research Institute Alexandria University, Egypt
| | - Samar Sayed
- Community Medicine Faculty of Medicine, Suez University, Egypt
| | - Manar Ezzelarab Ramadan
- Department of Medical Parasitology, Faculty of Medicine, Suez University, P.O. Box:43221, Suez, Egypt
| |
Collapse
|
4
|
Pliakou E, Lampropoulou D, Nasi D, Aravantinos G. Skin metastases from gastric cancer, a rare entity masquerading as erysipelas: A case report. Mol Clin Oncol 2022; 16:110. [DOI: 10.3892/mco.2022.2543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/06/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Evangelia Pliakou
- Second Department of Medical Oncology, General Oncology Hospital of Kifissia ‘Agioi Anargiroi’, Athens 14564, Greece
| | - Dimitra Lampropoulou
- Second Department of Medical Oncology, General Oncology Hospital of Kifissia ‘Agioi Anargiroi’, Athens 14564, Greece
| | - Despoina Nasi
- Second Department of Medical Oncology, General Oncology Hospital of Kifissia ‘Agioi Anargiroi’, Athens 14564, Greece
| | - Gerasimos Aravantinos
- Second Department of Medical Oncology, General Oncology Hospital of Kifissia ‘Agioi Anargiroi’, Athens 14564, Greece
| |
Collapse
|
5
|
Rasool KH, Mahmood Alubadi AE, Al-Bayati IFI. The role of Serum Interleukin-4 and Interleukin-6 in Helicobacter pylori-infected patients. Microb Pathog 2021; 162:105362. [PMID: 34942310 DOI: 10.1016/j.micpath.2021.105362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/04/2021] [Accepted: 12/12/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) is a Gram-negative bacteria that colonizes the gastrointestinal mucosa and causes chronic inflammation. AIMS There are several studies related to cytokines concentrations in H. pylori infection, but there is no study that clarifies the role of IL-4 and IL-6 together in H. pylori infection. SUBJECTS and methods: A total of (65) individuals, (25) healthy controls and (40) patients with H. pylori infection which are submitted to the hepato-gastrointestinal unit of AL-Imamian Al-Kadhimiyain medical hospital city in Baghdad consulted and diagnosed by traditional ways. In addition, the diagnosis was confirmed by ELISA Helicobacter pylori IgG kit. IL-4 and IL-6 levels were also assessed by using ELISA kits, according to the manufactures procedures. RESULTS Significant increase of IL-4 and IL-6 concentration in seropositive patients compared with seronegative controls. CONCLUSION Our findings and previous researches indicate that the immune response of gastric mucosa to H. pylori varies among patients. It depends on the stage of infection, and increases the cytokines secretion in the chronic stage, which is expected because of increased immune cell infiltration at the site of infection.
Collapse
Affiliation(s)
- Khetam Habeeb Rasool
- Department of Biology, College of Science, Mustansiriyah University, Baghdad, Iraq.
| | | | | |
Collapse
|
6
|
Wei Q, Gao Y, Qi C, Yuan X, Li J, Xu Q, Luo C, Chen L, Zhuo W, Xu Z, Ying J. Clinicopathological Characteristics and Prognosis of Signet Ring Gastric Cancer: A Population-Based Study. Front Oncol 2021; 11:580545. [PMID: 34490073 PMCID: PMC8418067 DOI: 10.3389/fonc.2021.580545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 06/16/2021] [Indexed: 12/14/2022] Open
Abstract
Background To better define the clinicopathologic characteristics of signet ring cell (SRC) gastric cancer and build a prognostic model for it. Methods SRC patient information from 2010 to 2015 were identified using Surveillance, Epidemiology, and End Results (SEER) database. Kaplan-Meier method and log-rank test were used to estimate Overall survival (OS) and to determine associations with histologic subtypes. In COX proportional hazards regression model–based univariate and multivariate analyses, significant variables for construction of a nomogram were screened out. The nomogram was validated by means of the concordance index (CI), calibration plots, and receiver operating characteristics (ROCs) curves. Results A total of 11,363 gastric cancer patients were enrolled. On dividing the patients into SRC, well-to-moderately differentiated (WMD) adenocarcinoma, and poorly differentiated (PD) adenocarcinoma, differences among these subgroups emerged. SRC patients were more likely to occur in female and young patients than other histologic subtypes. Larger tumors, stage T4, and node stage N3 were more likely to be found in the SRC group. The survival for SRC patients was better than non-SRC patients in stage I. Univariate and multivariate analyses identified age, tumor site, larger tumor size, advanced T classification, advanced N classification, advanced TNM stage, and surgery of primary site as independent prognostic indicators. Then an OS nomogram was formulated. Conclusions SRC had distinct clinicopathological characteristics. The nomogram provided an accurate tool to evaluate the prognosis of SRC.
Collapse
Affiliation(s)
- Qing Wei
- Department of Abdominal Medical Oncology, Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yiding Gao
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Changsong Qi
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xing Yuan
- Department of Abdominal Medical Oncology, Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jingjing Li
- Department of Abdominal Medical Oncology, Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Qi Xu
- Department of Abdominal Medical Oncology, Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Cong Luo
- Department of Abdominal Medical Oncology, Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lei Chen
- Department of Abdominal Medical Oncology, Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Wei Zhuo
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, Institute of Cancer and Basic Medicine (ICBM), Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, China
| | - Jieer Ying
- Department of Abdominal Medical Oncology, Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
7
|
Predictive value of m5C regulatory gene expression in pancreatic adenocarcinoma. Sci Rep 2021; 11:17529. [PMID: 34471186 PMCID: PMC8410865 DOI: 10.1038/s41598-021-96470-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/10/2021] [Indexed: 11/08/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is the most malignant digestive tumor. The global incidence of pancreatic cancer has been rapidly trending upwards, necessitating an exploration of potential prognostic biomarkers and mechanisms of disease development. One of the most prevalent RNA modifications is 5-methylcytosine (m5C); however, its contribution to PAAD remains unclear. Data from The Cancer Genome Atlas (TCGA) database, including genes, copy number variations (CNVs), and simple nucleotide variations (SNVs), were obtained in the present study to identify gene signatures and prognostic values for m5C regulators in PAAD. Regulatory gene m5C changes were significantly correlated with TP53, BRCA1, CDKN2A, and ATM genes, which play important roles in PAAD pathogenesis. In particular, there was a significant relationship between m5C regulatory gene CNVs, especially in genes encoding epigenetic “writers”. According to m5C-regulated gene expression in clinically graded cases, one m5C-regulated genes, DNMT3A, showed both a strong effect on CNVs and a significant correlation between expression level and clinical grade (P < 0.05). Furthermore, low DNMT3A expression was not only associated with poor PAAD patient prognosis but also with the ribosomal processing. The relationship between low DNMT3A expression and poor prognosis was confirmed in an International Cancer Genome Consortium (ICGC) validation dataset.
Collapse
|
8
|
Olesiński T, Lutkowska A, Balcerek A, Sowińska A, Piotrowski P, Trzeciak T, Maj T, Hevelke P, Jagodziński PP. Long noncoding RNA CCAT1 rs67085638 SNP contribution to the progression of gastric cancer in a Polish population. Sci Rep 2021; 11:15369. [PMID: 34321511 PMCID: PMC8319342 DOI: 10.1038/s41598-021-94576-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/07/2021] [Indexed: 01/17/2023] Open
Abstract
The role of the long noncoding RNA CCAT1 NC_000008.10:g.128220661C > T (rs67085638) in the development of colon cancer has been reported. Therefore, we assessed the prevalence of rs67085638 in patients with gastric cancer (GC). We also evaluated the effect of rs67085638 on B-cell-specific Moloney leukaemia virus insertion site 1 (BMI1) transcripts in primary GC and counterpart histopathologically confirmed disease-free margin tissue. Using high-resolution melting analysis, we evaluated rs67085638 frequency in patients with the GC genotype (n = 214) and controls (n = 502) in a Polish Caucasian population. qRT-PCR was used to determine BMI1 transcripts. We observed the trend of rs67085638 association in all patients with GC (ptrend = 0.028), a strong risk of the GC genotype in male (ptrend = 0.035) but not female (ptrend = 0.747) patients, and the association with non-cardia GC (ptrend = 0.041), tumour stages T3 (ptrend = 0.014) and T4 (ptrend = 0.032), differentiation grading G3 (ptrend = 0.009), lymph node metastasis stage N3 (ptrend = 0.0005) and metastasis stage M0 (ptrend = 0.027). We found that significantly increased BMI1 transcripts were associated with the primary GC genotype classified as grade G3 (p = 0.011) and as lymph node metastasis N3 (p = 0.010) and counterpart marginal tissues (p = 0.026, p = 0.040, respectively) from carriers of the T/T versus C/C genotypes. rs67085638 may contribute to increased BMI1 transcripts and the progression and rapid growth of GC.
Collapse
Affiliation(s)
- Tomasz Olesiński
- Department of Oncological Gastroenterology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Anna Lutkowska
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, 6 Święcickiego St., 60-781, Poznan, Poland
| | - Adam Balcerek
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, 6 Święcickiego St., 60-781, Poznan, Poland
| | - Anna Sowińska
- Department of Computer Science and Statistics, Poznań University of Medical Sciences, Poznan, Poland
| | - P Piotrowski
- Molecular Biology Department, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Tomasz Trzeciak
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Tomasz Maj
- Department of Oncological Gastroenterology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Piotr Hevelke
- Department of Oncological Gastroenterology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Pawel P Jagodziński
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, 6 Święcickiego St., 60-781, Poznan, Poland.
| |
Collapse
|
9
|
Identification and Verification of Molecular Subtypes with Enhanced Immune Infiltration Based on m6A Regulators in Cutaneous Melanoma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:2769689. [PMID: 33490266 PMCID: PMC7801086 DOI: 10.1155/2021/2769689] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/13/2020] [Accepted: 12/22/2020] [Indexed: 02/08/2023]
Abstract
Background As the most aggressive type of skin cancer, cutaneous melanoma (CM) is experiencing a rapidly rising mortality in recent years. Exploring potential prognostic biomarkers or mechanisms of disease progression therefore has a great significance for CM. The purpose of this study was to identify genetic markers and prognostic performance of N6-methyladenosine (m6A) regulators in CM. Method Gene expression profiles, copy number variation (CNV), and single nucleotide polymorphism (SNP) data of patients were obtained from The Cancer Genome Atlas (TCGA) database. Results Genomic variation and association analysis of gene expressions revealed a high degree of genomic variation in the presence of m6A-regulated genes. m6A patients with high-frequency genomic variants in the regulatory gene tended to develop a worse prognosis (p < 0.01). Unsupervised cluster analysis of the expression profiles of m6A-regulated genes identified three clinically distinct molecular subtypes, including degradation-enhanced subgroup and immune-enhanced subgroup, with significant prognostic differences (p = 0.046). A novel prognostic signature, which was established according to m6A-related characteristic genes identified through genome-wide expression spectrum, could effectively identify samples with poor prognosis and enhanced immune infiltration, and the effectiveness was also verified in the dataset of the chip. Conclusion We identified genetic changes in the m6A regulatory gene in CM and related survival outcomes. The findings of this study provide new insights into the epigenetic understanding of m6A in CM.
Collapse
|
10
|
Wang L, Li C, Tian J, Liu J, Zhao Y, Yi Y, Zhang Y, Han J, Pan C, Liu S, Deng N, Xian Z, Li G, Zhang X, Liang A. Genome-wide transcriptional analysis of Aristolochia manshuriensis induced gastric carcinoma. PHARMACEUTICAL BIOLOGY 2020; 58:98-106. [PMID: 31957525 PMCID: PMC7006638 DOI: 10.1080/13880209.2019.1710219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Context: Aristolochia manshuriensis Kom (Aristolochiaceae) (AMK) is known for toxicity and mutagenicity.Objective: The tumorigenic role of AMK has yet to be understood.Materials and methods: AMK extracts were extracted from root crude drug. SD (Sprague Dawley) rats underwent gavage with AMK (0.92 g/kg) every other day for 10 (AMK-10) or 20 (AMK-20) weeks. Stomach samples were gathered for histopathological evaluation, microarray and mRNA analysis.Results: The gastric weight to body weight ratio (GW/BW) is 1.7 in the AMK-10 cohort, and 1.8 in AMK-20 cohort compared to control (CTL) cohort. Liver function was damaged in AMK-10 and AMK-20 rats compared to CTL rats. There were no significant changes of CRE (creatinine) in AMK-10 and AMK-20 rats. Histopathological analysis revealed that rats developed dysplasia in the forestomach in AMK-10 rats, and became gastric carcinoma in AMK-20 rats. Genes including Mapk13, Nme1, Gsta4, Gstm1, Jun, Mgst2, Ggt6, Gpx2, Gpx8, Calml3, Rasgrp2, Cd44, Gsr, Dgkb, Rras, and Amt were found to be critical in AMK-10 and AMK-20 rats. Pik3cb, Plcb3, Tp53, Hras, Myc, Src, Akt1, Gnai3, and Fgfr3 worked in AMK-10 rats, and PDE2a and PDE3a played a pivotal role in AMK-20 rats.Discussion and conclusions: AMK induced benign or malignant gastric tumours depends on the period of AMK administration. Genes including Mapk13, Nme1, Gsta4, Gstm1, Jun, Mgst2, Ggt6, Gpx2, Gpx8, Calml3, Rasgrp2, Cd44, Gsr, Dgkb, Rras, Amt, Pik3cb, Plcb3, Tp53, Hras, Myc, Src, Akt1, Gnai3, Fgfr3, PDE2a, and PDE3a were found to be critical in aristolochic acid-induced gastric tumour process.
Collapse
Affiliation(s)
- Lianmei Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Chunying Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Jingzhuo Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Jing Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Yong Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Yan Yi
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Yushi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Jiayin Han
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Chen Pan
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Suyan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Nuo Deng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Zhong Xian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Guiqin Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Xin Zhang
- Blood Products Engineering Research and Development Center, Shenzhen, China
| | - Aihua Liang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
- CONTACT Aihua Liang Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| |
Collapse
|
11
|
Zhu J, Deng L, Chen B, Huang W, Lin X, Chen G, Tzeng CM, Ying M, Lu Z. Magnesium-dependent Phosphatase (MDP) 1 is a Potential Suppressor of Gastric Cancer. Curr Cancer Drug Targets 2020; 19:817-827. [PMID: 31218958 DOI: 10.2174/1568009619666190620112546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/19/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Recurrence is the leading cause of treatment failure and death in patients with gastric cancer (GC). However, the mechanism underlying GC recurrence remains unclear, and prognostic markers are still lacking. METHODS We analyzed DNA methylation profiles in gastric cancer cases with shorter survival (<1 year) or longer survival (> 3 years), and identified candidate genes associated with GC recurrence. Then, the biological effects of these genes on gastric cancer were studied. RESULTS A novel gene, magnesium-dependent phosphatase 1 (mdp1), was identified as a candidate gene whose DNA methylation was higher in GC samples from patients with shorter survival and lower in patients with longer survival. MDP1 protein was highly expressed in GC tissues with longer survival time, and also had a tendency to be expressed in highly differentiated GC samples. Forced expression of MDP1 in GC cell line BGC-823 inhibited cell proliferation, whereas the knockdown of MDP1 protein promoted cell growth. Overexpression of MDP1 in BGC-823 cells also enhanced cell senescence and apoptosis. Cytoplasmic kinase protein c-Jun N-terminal kinase (JNK) and signal transducer and activator of transcription 3 (Stat3) were found to mediate the biological function of MDP1. CONCLUSION These results suggest that MDP1 protein suppresses the survival of gastric cancer cells and loss of MDP expression may benefit the recurrence of gastric cancer.
Collapse
Affiliation(s)
- Jianbo Zhu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Lijuan Deng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Baozhen Chen
- Department of Pathology, Fujian Provincial Tumor Hospital, 420 Fuma Road, Fuzhou, Fujian 350014, China
| | - Wenqing Huang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Xiandong Lin
- Department of Pathology, Fujian Provincial Tumor Hospital, 420 Fuma Road, Fuzhou, Fujian 350014, China
| | - Gang Chen
- Department of Pathology, Fujian Provincial Tumor Hospital, 420 Fuma Road, Fuzhou, Fujian 350014, China
| | - Chi-Meng Tzeng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361005, China.,Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen, Fujian 361005, China
| | - Mingang Ying
- Department of Pathology, Fujian Provincial Tumor Hospital, 420 Fuma Road, Fuzhou, Fujian 350014, China
| | - Zhongxian Lu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361005, China.,Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen, Fujian 361005, China
| |
Collapse
|
12
|
Azevedo MM, Pina-Vaz C, Baltazar F. Microbes and Cancer: Friends or Faux? Int J Mol Sci 2020; 21:ijms21093115. [PMID: 32354115 PMCID: PMC7247677 DOI: 10.3390/ijms21093115] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/23/2020] [Accepted: 04/26/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer is one of the most aggressive and deadly diseases in the world, representing the second leading cause of death. It is a multifactorial disease, in which genetic alterations play a key role, but several environmental factors also contribute to its development and progression. Infections induced by certain viruses, bacteria, fungi and parasites constitute risk factors for cancer, being chronic infection associated to the development of certain types of cancer. On the other hand, susceptibility to infectious diseases is higher in cancer patients. The state of the host immune system plays a crucial role in the susceptibility to both infection and cancer. Importantly, immunosuppressive cancer treatments increase the risk of infection, by decreasing the host defenses. Furthermore, alterations in the host microbiota is also a key factor in the susceptibility to develop cancer. More recently, the identification of a tumor microbiota, in which bacteria establish a symbiotic relationship with cancer cells, opened a new area of research. There is evidence demonstrating that the interaction between bacteria and cancer cells can modulate the anticancer drug response and toxicity. The present review focuses on the interaction between microbes and cancer, specifically aiming to: (1) review the main infectious agents associated with development of cancer and the role of microbiota in cancer susceptibility; (2) highlight the higher vulnerability of cancer patients to acquire infectious diseases; (3) document the relationship between cancer cells and tissue microbiota; (4) describe the role of intratumoral bacteria in the response and toxicity to cancer therapy.
Collapse
Affiliation(s)
- Maria Manuel Azevedo
- Department of Microbiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- CINTESIS, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Agrupamento de Escolas D. Maria II, 4760-067 V.N. Famalicão, Portugal
- Correspondence: ; Tel.: +351-22-551-36
| | - Cidália Pina-Vaz
- Department of Microbiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- CINTESIS, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4835-258 Guimarães, Portugal
| |
Collapse
|
13
|
Ge S, Wang S, Xiang W, Wang L, Zhu Y, Zhu X, Wang X, Zuo L, Jiang C, Li S, Liu M. [Association of adenylate cyclase-associated protein 2 expression with histopathology and long-term prognosis of gastric cancer]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:1052-1058. [PMID: 31640951 DOI: 10.12122/j.issn.1673-4254.2019.09.08] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
OBJECTIVE To explore association of the expression levels of adenylate cyclase-associated protein 2 (CAP2) in gastric cancer tissues with the histopathology and long-term prognosis of the malignancy. METHODS This study was conducted among a total of 105 patients with gastric cancer undergoing radical gastrectomy in our hospital between January, 2010 and October, 2013. Immunohistochemistry was used to quantitatively assess the expression of CAP2 in gastric cancer tissues and the adjacent tissues. Based on the median relative expression level of CAP2 of 3.5, the patients were divided into low CAP2 expression group (n=52) and high CAP2 expression group (n=53). The Cox regression model was used to analyze the effect of CAP2 expression on the 5-year survival rate of the patients, and ROC curve analysis was used to assess the predictive value of CAP2 expression for the patients' long-term survival. RESULTS Immunohistochemical analysis showed that the expression levels of CAP2 (P < 0.01) and Ki67 (P < 0.01) were significantly higher in gastric cancer tissues than in the adjacent tissues, and the expression level of CAP2 was positively correlated with Ki67 (P < 0.01), peripheral blood CEA (P < 0.01) and CA19-9 (P < 0.01). The percentages of patients with CEA≥5 μg/L, CA19-9≥37 kU/L, pathological grade of G3-G4, T stage of 3-4, and N stage of 2-3 were significantly higher in patients with high CAP2 expression than in those with low CAP2 expression (P < 0.05). Kaplan- Meier survival analysis showed that the 5-year survival rate was significantly lower in patients with a high CAP2 expression (P < 0.01). A high expression level of CAP2, CEA≥5μg/L, CA19-9≥37 and pathological grades G3-G4 were all independent risk factors for shortened 5-year survival after radical gastrectomy (P < 0.01). With the relative expression level of 3.45 as the cut-off value, the sensitivity of CAP2 was 70.15% for predicting death 5 years after the surgery, with a specificity of 71.05% and an area under the curve of 0.779 (P < 0.01). CONCLUSIONS CAP2 is highly expressed in gastric cancer tissues in close relation with the tumor progression. CAP2 is an independent risk factor for 5-year survival rate after radical gastrectomy for gastric cancer and can be of clinical value in prognostic evaluation of the patients.
Collapse
Affiliation(s)
- Sitang Ge
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Shan Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Wujun Xiang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Lili Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Yuke Zhu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Xiang Zhu
- Department of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
| | - Xun Wang
- Department of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
| | - Lugen Zuo
- Department of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
| | - Congqiao Jiang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Siqing Li
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Mulin Liu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
14
|
Gao Y, Wang Y, Wang X, Wang Y, Zhang X, Sun X. TNF-like ligand 1A is associated with progression and prognosis of human gastric cancer. Onco Targets Ther 2019; 12:7715-7723. [PMID: 31571922 PMCID: PMC6756834 DOI: 10.2147/ott.s210939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose This study aimed to investigate the function of TNF-like ligand 1A (TL1A) in the tumorigenesis and progression of gastric cancer (GC). Methods RNA-seq gene expression and clinical information for GC patients were obtained from The Cancer Genome Atlas (TCGA) database. Differentially expressed genes (DEGs) between GC tissue samples and normal controls were screened with the edgeR package. Identification of gene co-expression and functional enrichment analyses were performed with Pearson’s correlation analysis and gene set enrichment analysis (GSEA), respectively. Lastly, survival analysis was performed using the Kaplan-Meier method with the log rank test. Results TL1A expression in GC tissue samples were significantly higher than that in normal controls (LogFC=1.07 and P=8.90E-07). Moreover, 215 genes, co-expressed with TL1A, and 21 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were obtained. Next, the miRNA-lncRNA/mRNA network, comprising 7 miRNAs, 27 lncRNAs, and 21 mRNAs, was constructed based on key genes from intersections between co-expression analysis and GSEA. In addition, survival analysis results demonstrated that TL1A (P=2.6e−07) was significantly associated with the overall survival (OS) of GC patients. Conclusion TL1A was involved in the tumorigenesis and progression of GC, and was significantly associated with the OS of GC patients.
Collapse
Affiliation(s)
- Yaxian Gao
- Department of Immunology, China Medical University, Shenyang, Liaoning 110000, People's Republic of China.,Department of Immunology, Chengde Medical College, Chengde, Hebei 067000, People's Republic of China
| | - Yuanyuan Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110000, People's Republic of China
| | - Xiao Wang
- Department of Immunology, China Medical University, Shenyang, Liaoning 110000, People's Republic of China
| | - Yongwei Wang
- Department of Anatomy, Chengde Medical College, Chengde, Hebei 067000, People's Republic of China
| | - Xiaoqing Zhang
- Department of Immunology, China Medical University, Shenyang, Liaoning 110000, People's Republic of China
| | - Xun Sun
- Department of Immunology, China Medical University, Shenyang, Liaoning 110000, People's Republic of China
| |
Collapse
|
15
|
Negovan A, Iancu M, Fülöp E, Bănescu C. Helicobacter pylori and cytokine gene variants as predictors of premalignant gastric lesions. World J Gastroenterol 2019; 25:4105-4124. [PMID: 31435167 PMCID: PMC6700706 DOI: 10.3748/wjg.v25.i30.4105] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/12/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer remains the third leading cause of mortality from cancer worldwide and carries a poor prognosis, due largely to late diagnosis. The importance of the interaction between Helicobacter pylori (H. pylori) infection, the main risk factor, and host-related genetic factors has been studied intensively in recent years. The genetic predisposition for non-hereditary gastric cancer is difficult to assess, as neither the real prevalence of premalignant gastric lesions in various populations nor the environmental risk factors for cancer progression are clearly defined. For non-cardiac intestinal-type cancer, identifying the factors that modulate the progression from inflammation toward cancer is crucial in order to develop preventive strategies. The role of cytokines and their gene variants has been questioned in regard to non-self-limiting H. pylori gastritis and its evolution to gastric atrophy and intestinal metaplasia; the literature now includes various and non-conclusive results on this topic. The influence of the majority of cytokine single nucleotide polymorphisms has been investigated for gastric cancer but not for preneoplastic gastric lesions. Among the investigated gene variants onlyIL10T-819C, IL-8-251, IL-18RAP917997, IL-22 rs1179251, IL1-B-511, IL1-B-3954, IL4R-398 and IL1RN were identified as predictors for premalignant gastric lesions risk. One of the most important limiting factors is the inhomogeneity of the studies (e.g., the lack of data on concomitant H. pylori infection, methods used to assess preneoplastic lesions, and source population). Testing the modifying effect of H. pylori infection upon the relationship between cytokine gene variants and premalignant gastric lesions, or even testing the interaction between H. pylori and cytokine gene variants in multivariable models adjusted for potential covariates, could increase generalizability of results.
Collapse
Affiliation(s)
- Anca Negovan
- Department of Clinical Science-Internal Medicine, University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Mureș 540139, Romania
| | - Mihaela Iancu
- Department of Medical Informatics and Biostatistics, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca, Cluj 400349, Romania
| | - Emőke Fülöp
- Department of Morphological Sciences, Histology, University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Mureș 540139, Romania
| | - Claudia Bănescu
- Genetics Laboratory, Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Mureș 540139, Romania
| |
Collapse
|
16
|
Zhu L, Chen W, Li G, Chen H, Liao W, Zhang L, Xiao X. Upregulated RACK1 attenuates gastric cancer cell growth and epithelial-mesenchymal transition via suppressing Wnt/β-catenin signaling. Onco Targets Ther 2019; 12:4795-4805. [PMID: 31417279 PMCID: PMC6592218 DOI: 10.2147/ott.s205869] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose: As there have been few studies on the effects of the receptor for activated C kinase 1 (RACK1) on gastric cancer (GC), we aimed to explore such effects and the mechanism that may be involved. Patients and methods: Normal gastric epithelial cells and six GC cell lines were used to detect the mRNA expression of RACK1. Overexpressing RACK1 was transfected in HGC27 and MGC803 cells. The effects of overexpressing RACK1 on cell viability, migration, and invasion were determined by cell counting kit-8, wound scratch, and Transwell assay, respectively. The expressions of epithelial–mesenchymal transition (EMT) and Wnt/β-catenin signaling related genes were detected using quantitative real-time PCR or Western blot. Wnt pathway agonist LiCl was added into RACK1 overexpressing GC cells, and then cell viability, migration, and invasion were also detected. Results: RACK1 was downregulated in GC cell lines. Under the circumstance that overexpressing RACK1 was successfully transfected in the two lowest RACK1-expressing GC cells, significant inhibition of cell viability, migration, and invasion, promotion to the mRNA and protein expression of E-cadherin, as well as a decrease in the N-cadherin and Snail expressions could be observed. Overexpressing RACK1 also enhanced the protein level of phosphorylation-β-catenin/β-catenin and attenuated c-Jun protein expression. Additionally, LiCl could partially reverse the inhibitory effects of cell viability, migration and invasion by overexpressing RACK. Conclusion: We found RACK1 possibly inhibited epithelial–mesenchymal transition of GC cells through limitation of the Wnt/β-catenin pathway, thereby suppressing cell migration and invasion; RACK1 could also suppress cell growth.
Collapse
Affiliation(s)
- Lihui Zhu
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Wen Chen
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Guoqing Li
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Honghui Chen
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Wenqiu Liao
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Li Zhang
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Xiaoli Xiao
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| |
Collapse
|
17
|
Dominguez RL, Cherry CB, Estevez-Ordonez D, Mera R, Escamilla V, Pawlita M, Waterboer T, Wilson KT, Peek RM, Tavera G, Williams SM, Gulley ML, Emch M, Morgan DR. Geospatial analyses identify regional hot spots of diffuse gastric cancer in rural Central America. BMC Cancer 2019; 19:545. [PMID: 31174492 PMCID: PMC6554991 DOI: 10.1186/s12885-019-5726-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/16/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Geospatial technology has facilitated the discovery of disease distributions and etiology and helped target prevention programs. Globally, gastric cancer is the leading infection-associated cancer, and third leading cause of cancer mortality worldwide, with marked geographic variation. Central and South America have a significant burden, particularly in the mountainous regions. In the context of an ongoing population-based case-control study in Central America, our aim was to examine the spatial epidemiology of gastric cancer subtypes and H. pylori virulence factors. METHODS Patients diagnosed with gastric cancer from 2002 to 2013 in western Honduras were identified in the prospective gastric cancer registry at the principal district hospital. Diagnosis was based on endoscopy and confirmatory histopathology. Geospatial methods were applied using the ArcGIS v10.3.1 and SaTScan v9.4.2 platforms to examine regional distributions of the gastric cancer histologic subtypes (Lauren classification), and the H. pylori CagA virulence factor. Getis-Ord-Gi hot spot and Discrete Poisson SaTScan statistics, respectively, were used to explore spatial clustering at the village level (30-50 rural households), with standardization by each village's population. H. pylori and CagA serologic status was determined using the novel H. pylori multiplex assay (DKFZ, Germany). RESULTS Three hundred seventy-eight incident cases met the inclusion criteria (mean age 63.7, male 66.3%). Areas of higher gastric cancer incidence were identified. Significant spatial clustering of diffuse histology adenocarcinoma was revealed both by the Getis-Ord-GI* hot spot analysis (P-value < 0.0015; range 0.00003-0.0014; 99%CI), and by the SaTScan statistic (P-value < 0.006; range 0.0026-0.0054). The intestinal subtype was randomly distributed. H. pylori CagA had significant spatial clustering only in association with the diffuse histology cancer hot spot (Getis-Ord-Gi* P value ≤0.001; range 0.0001-0.0010; SaTScan statistic P value 0.0085). In the diffuse gastric cancer hot spot, the lowest age quartile range was 21-46 years, significantly lower than the intestinal cancers (P = 0.024). CONCLUSIONS Geospatial methods have identified a significant cluster of incident diffuse type adenocarcinoma cases in rural Central America, suggest of a germline genetic association. Further genomic and geospatial analyses to identify potential spatial patterns of genetic, bacterial, and environmental risk factors may be informative.
Collapse
Affiliation(s)
| | - Charlotte B Cherry
- Office of Public Health Informatics & Analytics, Tennessee Department of Public Health, Nashville, TN, USA
| | - Dago Estevez-Ordonez
- Vanderbilt Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, USA
| | - Robertino Mera
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, USA
| | - Veronica Escamilla
- Carolina Population Center, University of North Carolina, Chapel Hill, USA
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Keith T Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, USA
| | - Richard M Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, USA
| | - Gloria Tavera
- Department of Population and Quantitative Health Sciences and Institute of Computational Biology, Case Western Reserve University, Cleveland, USA
| | - Scott M Williams
- Department of Population and Quantitative Health Sciences and Institute of Computational Biology, Case Western Reserve University, Cleveland, USA
| | - Margaret L Gulley
- Department of Pathology, University of North Carolina, Chapel Hill, USA
| | - Michael Emch
- Department of Geography, University of North Carolina, Chapel Hill, USA
| | - Douglas R Morgan
- Vanderbilt Ingram Cancer Center (VICC), Vanderbilt University Medical Center, Nashville, USA.
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, USA.
- Division of Gastroenterology and Hepatology, The University of Alabama at Birmingham (UAB), 1808 7th Avenue South, BDB 373, Birmingham, AL, 35233, USA.
| |
Collapse
|
18
|
Machlowska J, Pucułek M, Sitarz M, Terlecki P, Maciejewski R, Sitarz R. State of the art for gastric signet ring cell carcinoma: from classification, prognosis, and genomic characteristics to specified treatments. Cancer Manag Res 2019; 11:2151-2161. [PMID: 30936747 PMCID: PMC6421895 DOI: 10.2147/cmar.s188622] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is responsible for 9% of cancer deaths worldwide. Over 950,000 new cases are diagnosed each year, and about 90% of them are in advanced stage, requiring chemotherapy. In Europe there has been research based on pre- and postoperative chemotherapy treatment, using 5-fluorouracil, epirubicin, cisplatin, capecitabine, and docetaxel. Chemotherapy significantly impairs the quality of life of patients; however, the final effects are not always satisfactory. There is scientific evidence that gastric mucus tumors and signet ring cell carcinomas have a pattern of specific signatures, that distinguish them from other gastric cancer subtypes, and may be associated with a poor response to systematic treatment. Signet ring cell carcinoma is less chemosensitive than others, and the increase in the percentage of signet ring cells correlates with resistance to chemotherapy. Perioperative chemotherapy in advanced signet ring cell carcinomas is an independent factor of poor prognosis and survival, which is explained by the toxicity of neoadjuvant treatment. Therefore, curative surgical resection enhanced by standardized lymphadenectomy remains the recommended gold standard in GC therapy. According to presented studies, early detection and aggressive treatments for this subtype of GC is a reasonable approach. This review paper is mostly addressed to physicians who are interested in updating to the state of the art concerning different subtypes of gastric carcinoma.
Collapse
Affiliation(s)
- Julita Machlowska
- Department of Human Anatomy, Medical University of Lublin, Lublin, Poland,
| | - Małgorzata Pucułek
- Department of Human Anatomy, Medical University of Lublin, Lublin, Poland,
| | - Monika Sitarz
- Department of Conservative Dentistry and Endodontics, Medical University of Lublin, Lublin, Poland
| | - Paweł Terlecki
- Department of Surgery, St. John's Cancer Center, Lublin, Poland,
| | | | - Robert Sitarz
- Department of Human Anatomy, Medical University of Lublin, Lublin, Poland, .,Department of Surgery, St. John's Cancer Center, Lublin, Poland,
| |
Collapse
|
19
|
Calcagno DQ, Wisnieski F, Mota ERDS, Maia de Sousa SB, Costa da Silva JM, Leal MF, Gigek CO, Santos LC, Rasmussen LT, Assumpção PP, Burbano RR, Smith MAC. Role of histone acetylation in gastric cancer: implications of dietetic compounds and clinical perspectives. Epigenomics 2019; 11:349-362. [DOI: 10.2217/epi-2018-0081] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Histone modifications regulate the structural status of chromatin and thereby influence the transcriptional status of genes. These processes are controlled by the recruitment of different enzymes to a specific genomic site. Furthermore, obtaining an understanding of these mechanisms could help delineate alternative treatment and preventive strategies for cancer. For example, in gastric cancer, cholecalciferol, curcumin, resveratrol, quercetin, garcinol and sodium butyrate are natural regulators of acetylation and deacetylation enzyme activity that exert chemopreventive and anticancer effects. Here, we review the recent findings on histone acetylation in gastric cancer and discuss the effects of nutrients and bioactive compounds on histone acetylation and their potential role in the prevention and treatment of this type of cancer.
Collapse
Affiliation(s)
- Danielle Q Calcagno
- Programa de Pós-graduação em Oncologia e Ciências Médicas, Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, PA, Brazil
- Programa de Pós-graduação em Química Medicinal e Modelagem Molecular, Universidade Federal do Pará, Belém, PA, Brazil
- Residência Multiprofissional em Saúde/Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, PA, Brazil
| | | | - Elizangela R da Silva Mota
- Programa de Pós-graduação em Química Medicinal e Modelagem Molecular, Universidade Federal do Pará, Belém, PA, Brazil
| | - Stefanie B Maia de Sousa
- Programa de Pós-graduação em Oncologia e Ciências Médicas, Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, PA, Brazil
| | | | - Mariana F Leal
- Programa de Pós-graduação em Oncologia e Ciências Médicas, Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, PA, Brazil
- Disciplina de Genética, Universidade Federal de São Paulo, SP, Brazil
| | - Carolina O Gigek
- Disciplina de Genética, Universidade Federal de São Paulo, SP, Brazil
- Departamento de Patologia, Universidade Federal de São Paulo, SP, Brazil
| | - Leonardo C Santos
- Disciplina de Genética, Universidade Federal de São Paulo, SP, Brazil
| | - Lucas T Rasmussen
- Disciplina de Genética, Universidade Federal de São Paulo, SP, Brazil
- Pró-Reitoria de Pesquisa e Pós-Graduação, Universidade do Sagrado Coração, Bauru, SP, Brazil
| | - Paulo P Assumpção
- Programa de Pós-graduação em Oncologia e Ciências Médicas, Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, PA, Brazil
| | - Rommel R Burbano
- Programa de Pós-graduação em Oncologia e Ciências Médicas, Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, PA, Brazil
- Laboratório de Biologia Molecular, Hospital Ophir Loyola, Belém, PA, Brazil
| | - Marília AC Smith
- Disciplina de Genética, Universidade Federal de São Paulo, SP, Brazil
| |
Collapse
|
20
|
Wu Y, Tian S, Chen Y, Ji M, Qu Y, Hou P. miR-218 inhibits gastric tumorigenesis through regulating Bmi-1/Akt signaling pathway. Pathol Res Pract 2018; 215:243-250. [PMID: 30420101 DOI: 10.1016/j.prp.2018.10.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 10/14/2018] [Accepted: 10/26/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Previous studies indicated that miR-218 was deregulated in gastric cancer patients and correlated with tumor invasion and prognosis. The aim of this study was to clarify the effect of miR-218 on the malignant behavior of gastric cancer and its role in regulating Bmi-1/Akt signaling pathway. MATERIALS AND METHODS We used miR-218 mimic to transfect gastric cancer cell lines AGS and SGC-7901, and the overexpression efficiency was validated using qRT-PCR assay. MTT assay and Transwell chamber system were performed to detect the effect of miR-218 on cell proliferation, invasion and migration on gastric cancer. Western blot and qRT-PCR assay was used to test the role of miR-218 in regulating Bmi-1/Akt signaling pathway. RESULTS As shown in our research, ectopic expression of miR-218 in gastric cancer cells inhibits the proliferation, invasion and migration of gastric cancer cells. In addition, miR-218 re-expression inhibits the expression of Bmi-1 and its downstream target p-Akt473, as well as MMPs and EMT process. CONCLUSIONS miR-218 inhibits the proliferation, invasion and migration of gastric cancer cells through modulating EMT process and the expression of MMPs via Bmi-1/Akt signaling pathway.
Collapse
Affiliation(s)
- Yongxing Wu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Sijia Tian
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Yijun Chen
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Meiju Ji
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Yiping Qu
- Department of Radio-Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China.
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China.
| |
Collapse
|
21
|
Liu S, Liu JW, Sun LP, Gong YH, Xu Q, Jing JJ, Yuan Y. Association of IL10 gene promoter polymorphisms with risks of gastric cancer and atrophic gastritis. J Int Med Res 2018; 46:5155-5166. [PMID: 30205739 PMCID: PMC6300941 DOI: 10.1177/0300060518792785] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To investigate the association between polymorphisms of the interleukin 10 ( IL10) gene and risk of gastric cancer (GC) and atrophic gastritis (AG). METHODS This study enrolled patients with GC, patients with AG and healthy control subjects. Demographic data were collected and the IL10 gene -1082A/G, -819C/T and -592A/C polymorphisms were genotyped. An enzyme-linked immunosorbent assay was performed to detect Helicobacter pylori infection. RESULTS The study enrolled 556 participants including 208 in the GC group, 116 in the AG group and 232 controls (CON group). In a recessive model of the IL10-819C/T polymorphism, a significantly decreased risk of GC was found compared with AG and non-cancer subjects, respectively (AG→GC: odds ratio OR 0.41; non-cancer→GC: OR 0.57). The CC genotype demonstrated a significantly increased risk of AG compared with CON. Similar significant results were detected in males and H. pylori-negative subgroups. The ACC haplotype was associated with a decreased risk of GC compared with AG. The ATC haplotype was associated with a decreased risk of AG compared with the CON group, but it was associated with an increased risk of GC compared with AG. CONCLUSION The IL10 gene promoter -819C/T (rs1800871) polymorphism was associated with the risk of GC and AG in a Chinese population.
Collapse
Affiliation(s)
- Sa Liu
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China.,2 The Second Department of Oncology, Fourth Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jing-Wei Liu
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| | - Li-Ping Sun
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| | - Yue-Hua Gong
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| | - Qian Xu
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| | - Jing-Jing Jing
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| | - Yuan Yuan
- 1 Tumour Etiology and Screening Department of Cancer Institute and General Surgery, First Hospital of China Medical University, and Key Laboratory of Cancer Aetiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, Liaoning Province, China
| |
Collapse
|
22
|
Pucułek M, Machlowska J, Wierzbicki R, Baj J, Maciejewski R, Sitarz R. Helicobacter pylori associated factors in the development of gastric cancer with special reference to the early-onset subtype. Oncotarget 2018; 9:31146-31162. [PMID: 30123433 PMCID: PMC6089554 DOI: 10.18632/oncotarget.25757] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/22/2018] [Indexed: 02/07/2023] Open
Abstract
Nowadays, gastric cancer is one of the most common neoplasms and the fourth cause of cancer-related death on the world. Regarding the age at the diagnosis it is divided into early-onset gastric carcinoma (45 years or younger) and conventional gastric cancer (older than 45). Gastric carcinomas are rarely observed in young population and rely mostly on genetic factors, therefore provide the unique model to study genetic and environmental alternations. The latest research on early-onset gastric cancer are trying to explain molecular and genetic basis, because young patients are less exposed to environmental factors predisposing to cancer. In the general population, Helicobacter pylori, has been particularly associated with intestinal subtype of gastric cancers. The significant association of Helicobacter pylori infection in young patients with gastric cancers suggests that the bacterium has an etiologic role in both diffuse and intestinal subtypes of early-onset gastric cancers. In this paper we would like to ascertain the possible role of Helicobacter pylori infection in the development of gastric carcinoma in young patients. The review summarizes recent literature on early-onset gastric cancers with special reference to Helicobacter pylori infection.
Collapse
Affiliation(s)
| | | | - Ryszard Wierzbicki
- 2 Department of Surgery with Trauma, Orthopaedic and Urological Subunit, Independent Public Health Care Center of the Ministry of Interior and Administration in Lublin, Poland
- 3 Department of Surgical Oncology, Medical University of Lublin, Poland
| | - Jacek Baj
- 1 Department of Human Anatomy, Medical University of Lublin, Poland
| | | | - Robert Sitarz
- 1 Department of Human Anatomy, Medical University of Lublin, Poland
- 2 Department of Surgery with Trauma, Orthopaedic and Urological Subunit, Independent Public Health Care Center of the Ministry of Interior and Administration in Lublin, Poland
- 4 Department of Surgery, St. John's Cancer Center, Lublin, Poland
| |
Collapse
|
23
|
Zhan XY, Zhang Y, Zhai E, Zhu QY, He Y. Sorting nexin-1 is a candidate tumor suppressor and potential prognostic marker in gastric cancer. PeerJ 2018; 6:e4829. [PMID: 29868263 PMCID: PMC5983015 DOI: 10.7717/peerj.4829] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022] Open
Abstract
Sorting nexin-1 (SNX1) is an important functional protein in cell endocytosis, efflux, protein sorting, cell signal transduction, etc; however, the expression, the role and clinical relevance of SNX1 have not been investigated in gastric cancer (GC). In this study, we first performed a bioinformatics investigation using the data obtained from The Cancer Genome Atlas (TCGA) database. The result showed that SNX1 mRNA levels were significantly lower in GC tissues than in paracancerous tissues. In a study of 150 cases of GC, including 60 cases with paired paracancerous and cancer tissues and 90 cases with detailed follow-up information, SNX1 expression was analyzed by immunohistochemistry. Our study on paired paracancerous and cancer tissues showed that SNX1 protein expression remarkably decreased in GC tissues (50/60, 83.33%). A study on 90 patients with detailed follow-up information showed that tumors with higher SNX1 protein level were correlated with better clinicopathologic stages (p = 0.0285), nodal status (p = 0.0286), smaller tumor sizes (p = 0.0294) and a better survival rate in patients with GC (p = 0.0245). Univariate analysis of the 90 patients with GC showed that low-level SNX1 was significantly correlated with decreased overall survival of GC patients (p = 0.008), and associated with a relatively higher cumulative hazard of death. Exogenous expression of SNX1 inhibited the growth, migration, invasion and promoted the apoptosis and enhanced the sensitivity of GC cells to the chemotherapeutic drug 5-Fluorouracil (5-Fu) in vitro, while knockdown of SNX1 by short hairpin RNA (shRNA) significantly promoted the growth, migration, invasion and reduced the apoptosis and the sensitivity of GC cells to 5-Fu. SNX1 also showed to influence the levels of epithelial-mesenchymal transition markers including Vimentin, Snail, and E-cadherin in GC cells in vitro. Taken together, we propose here that SNX1 serves as a tumor suppressor and prognostic marker that reduces tumor cell malignancy for GC.
Collapse
Affiliation(s)
- Xiao-Yong Zhan
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangzhou KingMed Center for Clinical Laboratory, Guangzhou, China.,KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yaqiong Zhang
- Guangzhou KingMed Center for Clinical Laboratory, Guangzhou, China.,KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Ertao Zhai
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing-Yi Zhu
- Guangzhou KingMed Center for Clinical Laboratory, Guangzhou, China.,KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yulong He
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
24
|
Abstract
Elevated plasma fibrinogen levels and tumor progression in patients with gastric cancer (GC) have been largely reported. However, distinct fibrinogen chains and domains have different effects on coagulation, inflammation, and angiogenesis. The aim of this study was to characterize fibrinogen β chain (FGB) in GC tissues. Retrospectively we analyzed the data of matched pairs of normal (N) and malignant tissues (T) of 28 consecutive patients with GC at diagnosis by combining one- and two-dimensional electrophoresis (1DE and 2DE) with immunoblotting and mass spectrometry together with two-dimensional difference in gel electrophoresis (2D-DIGE). 1DE showed bands of the intact FGB at 50 kDa and the cleaved forms containing the fragment D at ~37–40 kDa, which corresponded to 19 spots in 2DE. In particular, spot 402 at ~50 kDa and spots 526 and 548 at ~37 kDa were of interest by showing an increased expression in tumor tissues. A higher content of spot 402 was associated with stomach antrum, while spots 526 and 548 amounts correlated with corpus and high platelet count (>208 × 109/L). The quantification of FGB and cleaved products may help to further characterize the interconnections between GC and platelet/coagulation pathways.
Collapse
|